AU2008290001B2 - Novel preparation for external use - Google Patents

Novel preparation for external use Download PDF

Info

Publication number
AU2008290001B2
AU2008290001B2 AU2008290001A AU2008290001A AU2008290001B2 AU 2008290001 B2 AU2008290001 B2 AU 2008290001B2 AU 2008290001 A AU2008290001 A AU 2008290001A AU 2008290001 A AU2008290001 A AU 2008290001A AU 2008290001 B2 AU2008290001 B2 AU 2008290001B2
Authority
AU
Australia
Prior art keywords
topical formulation
formulation according
compound
acid
bleeding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2008290001A
Other versions
AU2008290001A1 (en
Inventor
Chiharu Kimura
Maho Sakurai
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eisai R&D Management Co Ltd
Original Assignee
Eisai R&D Management Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eisai R&D Management Co Ltd filed Critical Eisai R&D Management Co Ltd
Publication of AU2008290001A1 publication Critical patent/AU2008290001A1/en
Application granted granted Critical
Publication of AU2008290001B2 publication Critical patent/AU2008290001B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/78Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 2
    • C07D239/84Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Cosmetics (AREA)

Abstract

A compound represented by the general formula (I), a salt of the compound, or a hydrate of either thereof. They have an excellent antipruritic effect and have an excellent metabolic effect. The compound represented by the general formula (I), salt of the compound, or hydrate of either thereof, in a preparation for external use, has excellent absorbability into the skin. The preparation for external use has excellent stability because the ingredients incorporated are less apt to separate out even in long-term storage. (I) [In the formula, R represents hydroxy, C alkoxy optionally substituted by C alkoxy, or amino optionally substituted by C alkyl.]

Description

FP08-0370-00 DESCRIPTION NOVEL PREPARATION FOR EXTERNAL USE Technical Field [0001] The present invention relates to a topical formulation comprising a 4-(3 5 benzoylaminophenyl)-6,7-dimethoxy-2-methylaminoquinazoline compound. Background Art [0002] Phosphodiesterase 4 inhibitor (hereinafter abbreviated as PDE4 inhibitor) is a drug that suppresses the action of the enzyme phosphodiesterase, which degrades cyclic AMP (hereinafter abbreviated as cAMP) and, as a result, has effects of 10 increasing intracellular cAMP concentrations to relax smooth muscle and suppressing activation of inflammatory cells. Therefore, the PDE4 inhibitor is used as a therapeutic agent for bronchial asthma, chronic obstructive pulmonary disease, allergic dermatitis such as atopic dermatitis and contact dermatitis, and the like. Example reports on PDE4 inhibitors are described below. Patent Document 1 15 discloses that 1,8-naphthyridine derivatives are effective for asthma. Patent Document 2 discloses an eye ointment containing roflumilast. Patent Document 3 discloses a method for treating inflammatory skin disease and allergic skin disease by locally administering a hydroxyindole compound. Patent Document 4 discloses a therapeutic agent for pruritus that contains a piperidine derivative. 20 [0003] A PDE4 inhibitor used for the treatment of an allergic dermatitis is often used as a topical product, which can be directly acted on the skin, in particular, an ointment. In general, an ointment is produced by adding a drug substance and ingredients such as an emulsifier, a solvent, a preservative, a moisturizing agent, and an absorption enhancer to an oily base or a water-soluble base, and mixing 25 these ingredients uniformly. However, some drugs may be hardly absorbed into the skin only by allowing these drugs to exist uniformly in the ointment. In such cases, a method of adding a absorption enhancer to the ointment or suspending or dissolving a drug in a water-soluble or lipid-soluble medium and then kneading the suspension or solution into a base with other additives. For example, Patent 30 Document 5 discloses an ointment produced by dissolving tacrolimus, which is an immunosuppressive agent, in propylene glycol by heating and mixing the solution with paraffin and petrolatum acting as bases, and isopropyl myristate acting as an absorption enhancer. Patent Document 6 discloses an ointment produced by C -\NRPonbADCCRBR\29923K3 I DOC-6/9/2I 1I dissolving flurbiprofen or indomethacin, non-steroidal anti-inflammatory agents, in 2 (2-methoxy-1-methylethyl)-5-methylcyclohexanol and mixing the solution with a petrolatum base. Patent Document 7 discloses a water-in-oil ointment produced by dissolving a drug such as azelastine hydrochloride, which is an anti-allergy drug, in 5 propylene glycol and mixing the solution with a white petrolatum base and isopropyl myristate or the like acting as an absorption enhancer. [0004] [Patent Document 1] Japanese Patent Laid-Open No. 2001-192385 [Patent Document 2] National Publication of International Patent Application No. 10 2005-529930 [Patent Document 3] National Publication of International Patent Application No. 2005-537262 [Patent Document 4] Japanese Patent Laid-Open No. 2005-47909 [Patent Document 5] Japanese Patent Laid-Open No. 5-17481 15 [Patent Document 6] Japanese Patent Laid-Open No. 8-165251 [Patent Document 7] Japanese Patent Laid-Open No. 2005-29541 [Patent Document 8] W099/37622 Disclosure of the Invention [0005] Some PDE4 inhibitors have a defect that they are poorly absorbed into the skin 20 when used as a topical product. Therefore, to improve the absorption property, it may occasionally be necessary to contain a liquid ingredient such as a large quantity of solvent for dissolving the compound or an absorption enhancer in the formulation. However, this may cause phase separation and bleeding of the liquid components from the formulation. In one or more aspects the present invention may advantageously 25 provide a topical formulation which allows high absorption of PDE4 inhibitor and does not result in bleeding of the component. [0006] As a result of intensive studies, the present inventors have found that a novel compound represented by the formula (I), salt thereof, or hydrate thereof is a PDE4 inhibitor that has an excellent anti-pruritic effect. 30 [Formula 1] 2 FP08-0370-00 0 H
H
3 CO N HaCO N N'CH3
H
3 COH H wherein R represents hydroxyl, CI- alkoxy optionally substituted with C 1
.
6 alkoxy, or amino optionally substituted with C 1
.
6 alkyl. [0007] The following compound is disclosed in Patent Document 8 as an example 5 of compounds that are similar to the compound represented by the formula () in structure. [Formula 2] H N
H
3 CO N
H
3 COJN NNH3 H It is described in Patent Document 8, that this compound has PDE4 10 inhibitory action and thereby has anti-inflammatory action based on the PDE4 inhibitory action. Although Patent Document 8 describes that the above compound is effective for the treatment of psoriasis based on the anti-inflammatory action, the above publication neither describes nor suggests application of the above compound to itch caused by atopic disease. Furthermore, Patent Document 8 15 neither describes nor suggests that the compound described in Patent Document 8 is effective for itch on which a steroid drug or an anti-histamine agent is not effective. In contrast, the present inventors have found that the compound represented by the formula (I), or salt thereof, or hydrate thereof has an excellent anti-pruritic effect and is effective for itch caused by atopic disease or the like. 20 [0008] Furthermore, the present inventors have found that the compound represented by the formula (1), or salt thereof or hydrate thereof has insufficient skin absorption properties when used as a topical formulation. Accordingly, as a result of further studies, the present inventors could developed the formulation that the absorption properties of the compound represented by the formula (I), salt 3 C:\NRPortblWCC\RBR\44461 _ I DOC-7/3/2012 thereof, or hydrate thereof can be improved, and the bleeding of the components from the formulation can be prevented, and then accomplished the present invention. [0009] The present invention provides the following [1] to [15]: [1] a topical formulation, comprising a compound represented by the formula (I), salt 5 thereof, or hydrate thereof, a solvent, a base and water: [Formula 3] S0 H N O.. - H3CO xj N
H
3 CO N' NCH3 H wherein R represents hydroxyl, CI.
6 alkoxy optionally substituted with CI.
6 alkoxy, or amino optionally substituted with CI.
6 alkyl. 10 [2] the topical formulation according to [1], wherein the compound represented by the formula (I) is methyl N- [3-(6,7-dimethoxy-2-methylaminoquinazolin-4 yl)phenyl]terephthalamic acid. [3] the topical formulation according to [1] or [2], comprising an absorption enhancer. [4] the topical formulation according to any one of [1] to [3], comprising a bleeding 15 preventing agent. [5] the topical formulation according to [4], comprising two or more types of bleeding preventing agents. [6] the topical formulation according to any one of [I] to [5], comprising water. [7] the topical formulation according to any one of [1] to [6], wherein the base is one 20 or more types selected from the group consisting of petrolatum, paraffin, liquid paraffin, microcrystalline wax, carunauba wax, and white beeswax. [8] the topical formulation according to any one of [1] to [7], wherein the solvent is one or more types selected from the group consisting of polyethylene glycol having a molecular weight of 200 to 600, dipropylene glycol, benzyl alcohol, polyoxyethylene 25 sorbitan fatty acid ester, diethylene glycol monoethyl ether, propylene glycol, polyoxyethylene oleyl ether, polyoxyethylene octyl phenyl ether, polyoxyethylene lauryl ether, polyoxyethylene castor oil, and oleic acid. [9] the topical formulation according to any one of [3] to [8], wherein the absorption 4 C 3NRPonbIDCCRBR\29923Hi. I DOC4/9/21II0 enhancer is one or more types selected from the group consisting of isopropyl myristate, ethyl myristate, octyldodecyl myristate, isopropyl palmitate, isostearyl palmitate, isopropyl isostearate, butyl stearate, ethyl oleate, decyl oleate, diisopropyl sebacate, diethyl sebacate, diisopropyl adipate, diethyl adipate, and diethyl phthalate. 5 [10] the topical formulation according to any one of [4] to [9], wherein the bleeding preventing agent is one or more types selected from the group consisting of polyethylene glycol having a molecular weight of 1000 to 50000, polyoxyethylene hydrogenated castor oil, stearic acid, oleic acid, sorbitan monostearate, sorbitan monooleate, sorbitan sesquioleate, sorbitan trioleate, and glycerol esters of fatty acids. 10 [11] the topical formulation according to [10], wherein the glycerol esters of fatty acids is one or more types selected from the group consisting of glyceryl monostearate, diglyceryl isostearate, and hexaglyceryl polyricinoleate. [12] the topical formulation according to [10], wherein the bleeding preventing agent is polyethylene glycol having a molecular weight of 1000 to 50000 and glycerol esters of 15 fatty acids. [13] the topical formulation according to 112], wherein the glycerol esters of fatty acids is glyceryl monostearate. [14] the topical formulation according to any one of [3] to [13], wherein the topical formulation comprises 10 to 30% by weight of a solvent and 5 to 20% by weight of an 20 absorption enhancer, and the sum of the solvent and absorption enhancer is 20 to 40% by weight based on the total preparation amount, respectively. [15] a method for preventing bleeding of liquid ingredients, comprising mixing polyethylene glycol having a molecular weight of 1000 to 50000 and glycerol esters of fatty acids in a topical formulation according to [1] to [14]. 25 Effect of the Invention [0010] The compound represented by the formula (I), salt thereof, or hydrate thereof of the present invention has an excellent anti-pruritic effect as well as an excellent effect in terms of metabolism. Furthermore, the topical formulation of the present invention has excellent skin absorption properties of the compound 5 FP08-0370-00 represented by the formula (I), salt thereof, or hydrate thereof. Furthermore, the topical formulation of the present invention has excellent stability because it can prevent the bleeding (the separation of the ingredients) the component from the formulation after storage over a long period of time. 5 Brief Description of the Drawings [0011] Figure 1 shows that dermatitis is suppressed by the ointment of the present invention. Figure 2 shows that scratching behaviors are suppressed by the ointment of the present invention. 10 Figure 3 is the powder X-ray diffraction patterns of the crystals obtained in Production example 25. Best Mode for Carrying Out the Invention [0012] The present invention will be described in detail below. [0013] In the present specification, the structural formula of a compound may 15 indicate a certain type of isomer, as a matter of convenience. The present invention includes all isomers generated because of the structure of a compound, such as a geometric isomer, an optical isomer, a stereoisomer, or a tautomer, and an isomeric mixture. Thus, the compound of the present invention is not limited to the descriptions of a formula provided as a matter of convenience, but it may be either 20 one of such isomers or a mixture thereof. Accordingly, an optically active form and a racemic form may exist in the compound of the present invention. In the present invention, such an optically active form and a racemic form are not limited, and any of them are included. In addition, a crystal polymorphism may also exist in the compound of the present invention. Such a crystal polymorphism is not 25 limited either, and the present invention may be either a single crystal form or a mixture thereof. Moreover, the present invention also includes an amorphous form, and the compound of the present invention includes an anhydrate and a hydrate. Furthermore, the present invention also includes so-called a metabolite, which is generated as a result of in vivo metabolism (oxidation, reduction, hydrolysis, 30 conjugation, etc.) of the compound (I) of the present invention. Still further, a compound (so-called a prodrug), which generates the compound (I) of the present invention as a result of in vivo metabolism (oxidation, reduction, hydrolysis, conjugation, etc.), is also included in the present invention. 6 FP08-0370-00 [0014] The definitions of terms, symbols, and others used in the present specification will be explained below, and the present invention will be described in detail below. [0015] The term "C.
6 alkyl" is used in the present specification to mean a linear or 5 branched-chain alkyl group containing 1 to 6 carbon atoms. Specific examples of
C
1
-
6 alkyl may include methyl, ethyl, 1-propyl (n-propyl), 2-propyl (i-propyl), 2 methyl-1-propyl (i-butyl), 2-methyl-2-propyl (t-butyl), 1-butyl (n-butyl), 2-butyl (s butyl), 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl 2-butyl, 3-methyl-2-butyl, 2,2,-dimethyl-1-propyl, 1-hexyl, 2-hexyl, 3-hexyl, 2 10 methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3 methyl-2-pentyl, 4-methyl-2-pentyl, 2-methyl-3-pentyl, 3-methyl-3-pentyl, 2,3 dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2,2-dimethyl-1-butyl, 2-ethyl-1-butyl, 3,3 dimethyl-2-butyl, and 2,3-dimethyl-2-butyl. Preferred examples may include C 1
.
3 alkyl such as methyl, ethyl, 1-propyl 15 (n-propyl), 2-propyl (i-propyl), 2-methyl-l-propyl (i-butyl), 2-methyl-2-propyl (t butyl), 1-butyl (n-butyl), or 2-butyl (s-butyl). More preferred examples may include methyl and ethyl. [0016] The term "C 1
.
6 alkoxy" is used in the present specification to mean an oxy group to which the above defined "C 1 .6 alkyl" binds. Specific examples of C 1 .6 20 alkoxy may include methoxy, ethoxy, 1-propoxy, 2-propoxy, 2-methyl-1-propoxy, 2-methyl-2-propoxy, 1-butoxy, 2-butoxy, 1-pentoxy, 2-pentyloxy, 3-pentyloxy, 2 methyl-1-butoxy, 3-methyl-1-butoxy, 2-methyl-2-butoxy, 3-methyl-2-butoxy, 2,2 dimethyl-1-propoxy, 1-hexyloxy, 2-hexyloxy, 3-hexyloxy, 2-methyl-1-pentoxy, 3 methyl-1-pentyloxy, 4-methyl-1-pentoxy, 2-methyl-2-pentoxy, 3-methyl-2-pentoxy, 25 4-methyl-2-pentoxy, 2-methyl-3-pentyloxy, 3-methyl-3-pentyloxy, 2,3-dimethyl-1 butoxy, 3,3-dimethyl-1-butoxy, 2,2-dimethyl-1-butoxy, 2-ethyl-1-butoxy, 3,3 dimethyl-2-butoxy, 2,3-dimethyl-2-butoxy and the like. Preferred examples may include C 1
.
3 alkoxy such as methoxy, ethoxy, 1 propoxy, and 2-propoxy. A more preferred example is methoxy. 30 In addition, examples of "C 1 .6 alkoxy optionally substituted with C 1 .6 alkoxy" in the definitions of R may include methoxymethoxy, ethoxymethoxy, methoxyethoxy, and ethoxyethoxy. [0017] Examples of "amino optionally substituted with C 1
.
6 alkyl" in the present 7 FP08-0370-00 specification may include amino, mono-CI.
6 alkylamino that is substituted with the aforementioned C 1 .6 alkyl (for example, methylamino, ethylamino, t-butylamino, etc.), di-C 1
.
6 alkylamino (for example, dimethylamino, diethylamino, methylethylamino, etc.) and the like. 5 Preferred examples may include amino, mono-C.
3 alkylamino, and di-C 1
.
3 alkylamino. More preferred examples may include amino and monomethylamino. [0018] The type of a "salt" used in the present specification is not particularly limited, as long as it forms a salt together with the compound of the present invention and it is pharmacologically acceptable. Examples of such a salt may 10 include an inorganic acid salt, an organic acid salt, an inorganic basic salt, an organic basic salt, an acidic or basic amino acid salt and the like. Preferred examples of an inorganic acid salt may include hydrochloride, hydrobromide, sulfate, nitrate, phosphate and the like. More preferred examples are hydrochloride, hydrobromide, sulfate, and phosphate. Preferred examples of an 15 organic acid salt may include acetate, succinate, fumarate, maleate, tartrate, citrate, lactate, stearate, benzoate, methanesulfonate, ethanesulfonate, p-toluenesulfonate, and benzenesulfonate and more preferred examples are methanesulfonate or p toluenesulfonate. Preferred examples of an inorganic basic salt may include: alkaline metal 20 salts such as a sodium salt or a potassium salt; alkaline-earth metal salts such as a calcium salt or a magnesium salt; aluminum salts; ammonium salts and the like. Preferred examples of an organic basic salt may include a diethylamine salt, a diethanolamine salt, a meglumine salt, an NN'-dibenzylethylenediamine salt and the like. 25 Preferred examples of an acidic amino acid salt may include aspartate and glutamate. Preferred examples of a basic amino acid salt may include an arginine salt, a lysine salt, an ornithine salt and the like. [0019] The topical formulation according to the present invention means an ointment preparation, a gel preparation, a cream preparation, a patch preparation, 30 an eye ointment preparation, a suppository preparation and the like and preferably an ointment preparation. [0020] The active ingredient in the topical formulation according to the present invention is a compound represented by the formula (I), salt thereof, or hydrate 8 FP08-0370-00 thereof: [Formula 4] H R -0
H
3 CO N HCO 'H3 H wherein R represents hydroxyl, C 1 . alkoxy optionally substituted with C 1
.
6 alkoxy, 5 or amino optionally substituted with C 1 . alkyl. [0021] Examples of the compound represented by the formula (I) include: methyl N-[ 3
-(
6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid; N-[3-( 6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid 10 hydrochloride; N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N',N' dimethylterephthalamide; ethyl N-[ 3
-(
6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid; 15 N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N' methylterephthalamide; propyl N-[ 3
-(
6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid; isopropyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 20 yl)phenyl]terephthalamic acid; N-[3-( 6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl]-N'-ethylterephthalamide; N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N' propylterephthalamide; N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N' 25 isopropylterephthalamide; methyl N-[ 3
-(
6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]isophthalic acid;
N-[
3
-(
6
,
7 -dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]isophthalic acid; ethyl N-[ 3
-(
6 ,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]isophthalic acid; 9 FP08-0370-00 propyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]isophthalic acid; isopropyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]isophthalic acid; 5 N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N methylisophthalamide; N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N ethylisophthalamide; N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N 10 propylisophthalamide N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]-N isopropylisophthalamide; N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid 2 methoxyethyl ester; and 15 N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]isophthalamic acid 2 methoxyethyl ester. Preferably, the compound represented by the formula (I) is methyl N-[3 (6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid. [0022] The compound represented by the formula (I) can be produced, for example, 20 by the method described below. [0023] [Formula 5] 0
NH
2 N OR'
H
3 CO , N CI (B-2) H 3 CO N
H
3 CO N N3CH 3
H
3 CO N N-CH3 H H (A-3) [wherein R1 represents C 1 , alkyl.] This is a method of allowing a compound (A-3) to react with a compound 25 (B-2) that is acid chloride in an inert solvent in the presence or absence of a base, so as to produce the compound (I-1) of the present invention. A compound (A-3) can be produced by production example 7 in WO 10 FP08-0370-00 99/37622. As such a compound (B-2), a known compound, a commercially available compound, or a compound that can easily be produced from a commercially available compound by a method that is generally carried out by those skilled in the 5 art, can be used. Examples of such a compound (B-2) may include 4 chlorocarbonyl benzoic acid methyl ester and the like. The compound (B-2) can be used in an amount of 1 to 10 times, and preferably 1 to 2 times the molar equivalent of the compound (A-3). The type of a solvent used is not particularly limited, as long as it dissolves 10 starting substances to a certain extent and it does not inhibit the reaction in the present step. Examples of a solvent may include: aromatic hydrocarbons such as toluene, benzene, xylene and the like; ethers such as diethyl ether, tetrahydrofuran, dimethoxyethane, dioxane and the like; halogenated hydrocarbons such as dichloromethane, chloroform, 1,2-dichloroethane, or carbon tetrachloride; organic 15 bases such as pyridine, 2-, 3- or 4-picoline and the like; water; and a mixture of these solvents. Preferred examples are tetrahydrofuran or pyridine. The type of a base used herein is not particularly limited, as long as a compound of interest can be obtained and non-separable by-products are not generated. Examples of a base may include: inorganic bases such as sodium 20 carbonate, potassium carbonate, sodium hydrogencarbonate, potassium hydrogencarbonate, cesium carbonate and the like; and organic bases such as pyridine, triethylamine and the like. A preferred example is pyridine. The aforementioned base can be used in an amount of 1 to 10 times, and preferably 1 to 4 times the molar equivalent of the compound (A-3). 25 The reaction temperature depends on a solvent and a reagent used. It is generally between -30*C and 180*C, and preferably between 0*C and 100*C. The reaction time depends on a solvent used and the reaction temperature. It is generally between 0.5 and 200 hours, and preferably between 1 and 100 hours. [0024] The compound represented by the formula (I) can be obtained by 30 hydrolyzing and esterifying or amidating the compound (I-1) if necessary. When the compound represented by the formula (I) is obtained in a free form, such a free form can be converted to a salt or hydrate according to common methods. Furthermore, when the compound represented by the formula (I) is obtained in a 11 FP08-0370-00 form of a salt or hydrate, these compounds can be converted to a free form according to common methods. [0025] The topical formulation of the present invention comprises a solvent and a base in addition to the aforementioned active ingredient. The present inventors 5 have found that, when a topical product is formulated by mixing an active ingredient and a base, skin absorption properties may become insufficient. The skin absorbability of the active ingredient in the topical formulation of the present invention is improved by adding a solvent. [0026] A solvent commonly used for a topical formulation can be used as the 10 solvent, and specific examples may include polyethylene glycol having a molecular weight of 200 to 600, dipropylene glycol, benzyl alcohol, polyoxyethylene sorbitan fatty acid ester, diethylene glycol monoethyl ether, propylene glycol, polyoxyethylene oleyl ether, polyoxyethylene octyl phenyl ether, polyoxyethylene lauryl ether, polyoxyethylene castor oil, and oleic acid. A preferred example of the 15 solvent is polyethylene glycol having a molecular weight of 200 to 600. Polyethylene glycol having a molecular weight of 200 to 600 means polyethylene glycol having an average molecular weight of 200 to 600 as a result of average molecular weight testing directed in the section of macrogol 400 in The Japanese Pharmacopoeia Fifteenth Edition. Among polyethylene glycols having a 20 molecular weight 200 to 600, polyethylene glycols 400 having an average molecular weight of 380 to 420 as a result of average molecular weight testing directed in the section of macrogol 400 in The Japanese Pharmacopoeia Fifteenth Edition are particularly preferred. [0027] A base commonly used as a base of a topical formulation, in particular, an 25 oleaginous base, can be used, and specific examples may include petrolatum, squalane, paraffin, liquid paraffin, microcrystalline wax, carunauba wax, white beeswax and the like. A preferred example of the base is petrolatum, and a particularly preferred example of the base is white petrolatum. [0028] The topical formulation of the present invention may further comprise an 30 absorption enhancer and/or bleeding preventing agent. The skin absorbability of the active ingredient can be further improved by adding an absorption enhancer. Furthermore, the bleeding of ingredients (in particular, a solvent and absorption enhancer) from the topical formulation of the present invention can be prevented 12 FP08-0370-00 by adding a bleeding preventing agent, and thus stability can be achieved. [0029] An absorption enhancer commonly used as an absorption enhancer of a topical formulation can be used, and specific examples may include isopropyl myristate, ethyl myristate, octyldodecyl myristate, isopropyl palmitate, isostearyl 5 palmitate, isopropyl isostearate, butyl stearate, ethyl oleate, decyl oleate, diisopropyl sebacate, diethyl sebacate, diisopropyl adipate, diethyl adipate, diethyl phthalate and the like. A preferred example of the absorption enhancer is isopropyl myristate. [0030] A bleeding preventing agent commonly used as a bleeding preventing agent 10 of a topical formulation can be used, and specific examples may include polyethylene glycol having a molecular weight of 1000 to 50000, polyoxyethylene hydrogenated castor oil, stearic acid, oleic acid, sorbitan monostearate, sorbitan monooleate, sorbitan sesquioleate, sorbitan trioleate, glycerol esters of fatty acids and the like. Examples of the glycerol esters of fatty acids may include glyceryl 15 monostearate, diglyceryl isostearate, hexaglyceryl polyricinoleate and the like, and a preferred example is glyceryl monostearate. It is preferable to use two or more types of bleeding preventing agents because the bleeding preventing effect can be increased. Preferred bleeding preventing agents are a combination of polyethylene glycol having a molecular 20 weight of 1000 to 50000 and glyceryl monostearate. Bleeding of a solvent, in particular, polyethylene glycol having a molecular weight of 200 to 600 can be prevented by using polyethylene glycol having a molecular weight of 1000 to 50000. Furthermore, bleeding of an absorption enhancer, in particular, isopropyl myristate can be prevented by using glyceryl monostearate. 25 The polyethylene glycol having a molecular weight 1000 to 50000 means polyethylene glycol having an average molecular weight of 1000 to 50000 as a result of average molecular weight testing directed in the section of macrogol 4000 in The Japanese Pharmacopoeia Fifteenth Edition. Among polyethylene glycols having a molecular weight of 1000 to 50000, polyethylene glycols 4000 having an 30 average molecular weight of 2600 to 3800 as a result of average molecular weight testing directed in the section of macrogol 4000 in The Japanese Pharmacopoeia Fifteenth Edition are particularly preferred. [0031] The topical formulation of the present invention may further comprise 13 FP08-0370-00 water. Degradation of an active ingredient can be suppressed by adding water. [0032] The topical formulation of the present invention may comprise a coloring agent, a flavoring agent, a preservative, an antioxidant, a stabilizer, a usability improving agent and the like other than the above ingredients. 5 [0033] The coloring agent may include iron sesquioxide, yellow iron sesquioxide, carmine, caramel, beta-carotene, titanium oxide, talc, riboflavin sodium phosphate, yellow aluminum lake, and the like. [0034] The flavoring agent may include cocoa powder, mentha oil, menthol, lemon oil, borneol, powdered cinnamon bark, ascorbic acid, citric acid, tartaric acid, 10 malic acid, aspartame, potassium acesulfame, and the like [0035]The preservative may include methylparaben, propylparaben, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid and the like. [0036] The antioxidant may include sulfite salts, ascorbic acid, tocopherol and the like. 15 [0037] The stabilizer may include ascorbic acid, edetic acid salt, erythorbic acid, tocopherol and the like. [0038] The usability improving agent may include polyoxyethylene hydrogenated castor oil 40, polyoxyethylene hydrogenated castor oil 60 and the like. [0039] The amounts of ingredients in the topical formulation of the present 20 invention can be suitably set, as long as the effect of the present invention is not impaired, but are preferably within the following ranges (expressed with % by weight based on the total amount of the formulation): active ingredient, 0.001 to 0.5% by weight; solvent, 10 to 30% by weight; base, 40 to 70% by weight; absorption enhancer, 5 to 20% by weight; bleeding preventing agent, 10 to 25% by 25 weight; and water, 0.1 to 5%. Here, the total amount of a solvent and an absorption enhancer is preferably 20 to 40% by weight. Furthermore, the amount of water mixed is preferably 0.3 to 3%, more preferably 0.5 to 2%. [0040] The topical formulation of the present invention can be manufactured according to common manufacturing methods for a topical formulation. Such a 30 method will be described below using an ointment as an example. First, the compound represented by the formula (I), salt thereof, or hydrate thereof, which is an active ingredient, is dissolved in a solvent by heating at 70*C to 80*C (Solution I). Meanwhile, an absorption enhancer and a bleeding preventing agent and other 14 FP08-0370-00 ingredients are added to the base if necessary and are dissolved by heating at 70*C to 80*C. And then, solution I and water if necessary are added to the resulting mixture, and the mixture is stirred at 70*C to 80*C for approximately 3 minutes. The mixing is maintained until the mixture was cooled down to approximately 5 32*C (around the human skin surface temperature) and an ointment is completed. An anti-oxidizing agent may be added to the solvent if necessary. Examples [0041] The compound represented by the formula (I), salt thereof, or hydrate thereof can be produced by the methods described in the following production 10 examples. However, these examples are provided for illustrative purposes only. Specific examples as described below are not intended to limit the scope of the invention in any case. In addition, various modifications may also be made within the scope of the present invention. Compounds, to which publication names or the like are attached, were produced in accordance with the publications or the like. 15 [0042] Production example A Synthesis of 3-(2-chloro-6,7-dimethoxy-quinazolin-4-yl)phenylamine [Formula 6]
NH
2
H
3 CO N
H
3 CO N CI Twenty-five grams of 2,4-dichloro-6,7-dimethoxyquinazoline was 20 suspended in 2.25 L of a mixed solution consisting of toluene:tetrahydrofuran:a 2 N sodium carbonate solution = 1:1:1. To the reaction mixture was added 21.5 g of 3-aminophenyl boronic acid 1/2 sulfate, and the mixture was degassed, the atmosphere in the reaction vessel was replaced with nitrogen. To the reaction mixture was added 2.23 g of tetrakis(triphenylphosphine)palladium(O), followed by 25 stirring at 60*C under a nitrogen atmosphere. Eighteen hours after initiation of the reaction, 1.2 g of tetrakis(triphenylphosphine)palladium(0) was added to the reaction mixture, and the stirring was continued. Thirty hours later, 1.2 g of 15 FP08-0370-00 tetrakis(triphenylphosphine)palladium(O) was further added to the reaction mixture, and stirring was further continued. Forty-eight hours after initiation of the reaction, the reaction mixture was cooled, and it was then transferred into a separatory funnel, so as to separate an organic layer. The obtained organic layer was washed 5 with 300 mL of brine, and was then dried over anhydrous magnesium sulfate. The desiccant was removed by passing it through 250 g of silica gel. The silica gel was washed with 1.5 L of ethyl acetate, and the obtained organic layers were combined and concentrated to dryness. The residue was triturated with 200 mL of ethyl acetate, and the obtained solid was then filtrated. The solid was washed with 100 10 mL of diethyl ether and 200 mL of a mixed solution consisting of n-heptane:ethyl acetate = 1:1, and dried under aeration to give 28.2 g of a target product. Yield: 92.5% 'H-NMR (DMSO-d 6 ) 8 (ppm): 3.86 (3H, s), 4.01 (3H, s), 5.40 (2H, br), 6.79 (1H, dd, J = 1.6, 8.0 Hz), 6.93 (1H, brd, J = 8.0 Hz), 7.02 (1H, t, J = 1.6 Hz), 7.24 (1H, t, 15 J = 8.0 Hz), 7.41 (1H, s), 7.43 (1H, s). [0043] Production example B Synthesis of [4-(3-aminophenyl)-6,7-dimethoxyquinazolin-2-yllmethylamine [Formula 7]
NH
2
H
3 CO N jk I 'iCH 3
H
3 CO N N H 20 Fourteen grams of 3-(2-chloro-6,7-dimethoxyquinazolin-4-yl)phenylamine was suspended in 135 mL of a mixed solution consisting of tetrahydrofuran:isopropanol = 2:1. To the reaction mixture was added 89 mL of a methylamine solution in methanol, and the reaction mixture was stirred in a pressure-resistant sealed tube reactor at 130*C for 24 hours. After the reaction 25 mixture was allowed to cool down to room temperature, it was diluted with 300 mL of ethyl acetate and then washed with 300 mL of water. A water layer was 16 FP08-0370-00 extracted with 100 mL of ethyl acetate, and the combined organic layer was washed with 100 mL of brine. The organic layer was separated and was then dried over anhydrous magnesium sulfate. The desiccant was removed by filtration, the organic layer was concentrated to dryness, and the resultant was triturated with a 5 mixed solvent consisting of ethyl acetate:tetrahydrofuran = 3:1. The obtained solid was filtrated, and the filtrate was then washed with ethyl acetate, and dried under aeration to yield 10 g of a product of interest. The filtrate was adsorbed on a 50 g silica gel column, and it was then eluted with a mixed solution consisting of ethyl acetate:methanol = 9:1, and the eluent was concentrated to dryness. The residue 10 was triturated with ethyl acetate, and the obtained solid was then filtrated. The solid was washed with diethyl ether, and dried under aeration to give 1.4 g of a target product. Total yield: 82.9% 1 H-NMR (CDCl 3 ) 5 (ppm): 3.12 (3H, d, J = 5.2 Hz), 3.80 (2H, brs), 3.82 (3H, s), 4.03 (3H, s), 5.30 (1H,br), 6.83 (IH, dd, J = 1.6, 8.0 Hz), 6.99 (1H, t, J = 1.6 Hz), 15 7.04 (1H, brd, J= 8.0 Hz), 7.07 (1H, s), 7.15 (1H, s), 7.30 (1H, t, J = 8.0 Hz). [0044] Production example C Alternative method for synthesis of 3-(2-chloro-6,7-dimethoxy-quinazolin-4 yl)phenylamine (production example A) To 634 g of sodium carbonate (5.98 mol) was added 2.91 kg of water under 20 a nitrogen atmosphere, followed by stirring for dissolution. To the solution were added 3.0 L of tetrahydrofuran, 431 g of 3-aminophenyl boronic acid monohydrate (2.78 mol), 30.4g of triphenylphosphine (0.116 mol) and 26.0 g of dichloropalladium (0.116 mol) in this order. To the mixture was dropwise added a solution of 2,4-dichloro-6,7-dimethoxyquinazoline (600 g; 2.32 mol) in 25 tetrahydrofuran (12.0 L) over 2 hours while stirring at 60*C, followed by stirring at the same temperature for 16 hours. To the mixture were added 3.0 kg of a 5% sodium chloride solution and 12.0 L of tetrahydrofuran in this order, and the mixture was stirred at 50*C for 1 hour and allowed to cool down to 25*C. The mixture was filtered through celite to remove the insoluble matter, and the filtrate 30 was transferred to separatory funnel and the organic layer was separated. To the separated organic layer were added 150 g of anhydrous magnesium sulfate and 60.0 g of activated carbon, and the mixture was stirred at 50*C for 1 hour and allowed to cool down to 25*C. The mixture was filtered through celite to remove 17 FP08-0370-00 the insoluble matter, and the filtrate was concentrated under reduced pressure. To the residue was added 6.0 L of water, and the mixture was stirred at room temperature for 1 hour, and precipitated crystals were collected by filtration. The collected crystals were dried at 50*C under reduced pressure to give 730 g (content 5 rate 62.2%) of a target product. Yield: 62.1% [0045] Production example D Alternative method for synthesis of [4-(3-aminophenyl)-6.7-dimethoxvquinazolin 2-yllmethylamine (production example B) Two hundred grams of crude 3-(2-chloro-6,7-dimethoxyquinazolin-4 10 yl)phenylamine (content 124 g; 0.394 mol) was suspended in a mixed solution consisting of 1.2L of tetrahydrofuran and 0.6 L of isopropanol. To the mixture was added 1.2 L of a methylamine solution in methanol, and the mixture was stirred in a SUS autoclave at 90*C for 15 hours. The reaction mixture was allowed to cool down to 25'C, and concentrated under reduced pressure. To the residue were 15 added 1.0 L of water and 4.0 L of chloroform, and the mixture was stirred at 50*C for 0.5 hours and allowed to cool down to 25*C. The mixture was filtered through celite to remove the insoluble matter, and the filtrate was transferred to separatory funnel and the organic layer was separated. To the separated organic layer were added 50.0 g of anhydrous magnesium sulfate and 20.0 g of activated carbon, and 20 the mixture was stirred at 50*C for 1 hour and allowed to cool down to 25*C. The mixture was filtered through celite to remove the insoluble matter, and the filtrate was concentrated under reduced pressure. To the residue was added 904 mL of chloroform, and the mixture was stirred at 50*C for 1 hour and stirred overnight after turning off the heater. Then the mixture was stirred in an ice bath for 2 hours 25 and precipitated crystals were collected by filtration. The collected crystals were dried at 50*C under reduced pressure to give 76.3 g of a target product. Yield: 38.7% [0046] Production example 1 Synthesis of methyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 30 yl)phenyllterephthalamic acid [Formula 8] 18 FP08-0370-00 0 H OCH 3 N 0
H
3 CO N K- (I CH 3
H
3 CO N H3 To a solution of 16.8 g of [4-(3-aminophenyl)-6,7-dimethoxyquinazolin-2 yl]methylamine and 8.6 g of pyridine dissolved in 300 mL of tetrahydrofuran was added 11.8 g of 4-chlorocarbonylbenzoic acid methyl ester at room temperature, 5 followed by stirring for 24 hours. To the reaction mixture was added 100 mL of dimethyl sulfoxide, the mixture was partitioned between a mixed solvent consisting of 2,000 mL of ethyl acetate and 1,000 mL of tetrahydrofuran, and 1,000 mL of a saturated sodium hydrogencarbonate solution, and the organic layer was separated. The water layer was further extracted with a mixed solvent consisting of 500 mL of 10 ethyl acetate and 500 mL of tetrahydrofuran. The combined organic layer was then washed with 1,000 mL of a saturated sodium hydrogencarbonate solution and 1,000 mL of brine in this order, and dried over anhydrous magnesium sulfate. The desiccant was removed by filtration with 100 g of a basic silica gel pad, followed by well washing with 2,000 mL of ethyl acetate. The combined eluent was 15 concentrated under reduced pressure, and the obtained crude product was suspended and triturated in a mixed solvent consisting of 100 mL of tetrahydrofuran and 500 mL of diethyl ether. The precipitated crystals were collected by filtration, washed twice with 100 niL of diethyl ether, and dried under aeration at 50*C for 5 hours to give 13.8 g of the crystals of the titled compound 20 (yield: 53.2%). 'H-NMR (DMSO-d 6 ) 8 (ppm): 2.88 (3H, d, J = 4.4 Hz), 3.74 (3H, s), 3.89 (3H, s), 3.92 (3H, s), 6.99 (1H, s), 7.00 (1H, brs), 7.17 (1H, s), 7.46 (1H, d, J = 8.0 Hz), 7.55 (lH, t, J = 8.0 Hz), 7.87 (1H, brd, J = 8.0 Hz), 8.08 (4H, s), 8.20 (1H, brs), 10.61 (1H, s). 25 [0047] Production example 2 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 19 FP08-0370-00 yl)phenyllterephthalamic acid hydrochloride [Formula 9] 0 OH H N 0 HCl
H
3 CO N
H
3 CO N N H To a solution of 2.5 g of methyl N-[3-(6,7-dimethoxy-2 5 methylaminoquinazolin-4-yl)phenyl]terephthalamic acid dissolved in a mixed solvent consisting of 50 mL of tetrahydrofuran and 25 mL of methanol was added 11.3 mL of a 5 N sodium hydroxide solution, followed by stirring at room temperature for 12 hours. The reaction mixture was adjusted to be acidic by addition of 5 N hydrochloric acid, and the obtained solid was then filtrated, washed 10 with 10 mL of water and 20 mL of ether, and dried under aeration to give 2.5 g of a target product. Yield: 95.3%. 'H-NMR (DMSO-d 6 ) 5 (ppm): 3.05 (3H, brs), 3.82 (3H, s), 3.98 (3H, s), 7.32 (1H, s), 7.54 (1H, brd, J = 8.0 Hz), 7.55 (1H, brs), 7.61 (1H, t, J = 8.0 Hz), 7.91 (1H, d, J = 8.0 Hz), 8.06 (4H, s), 8.35 (1H, brs), 10.71 (1H, s). 15 [0048] Production example 3 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N',N' dimethylterephthalamide [Formula 10] 20 FP08-0370-00 0 N'CH3 H I N
CH
3 0
H
3 CO N N
ACH
3
H
3 CO N HN H To a solution of 100 mg of N-(3-(6,7-dimethoxy-2-methylaminoquinazolin 4-yl)phenyl]terephthalamic acid hydrochloride dissolved in 2 mL of dimethylformamide were added 60 mg of WSC, 41 mg of 1-hydroxybenzotriazole, 5 42 pL of triethylamine, and 10 mg of 4-dimethylaminopyridine, followed by stirring at room temperature for 30 minutes. To the reaction mixture was added 200 pL of a dimethylamine solution in tetrahydrofuran, followed by stirring at room temperature for 15 hours. To the reaction mixture was added 2 mL of tetrahydrofuran, and the reaction mixture was partitioned after addition of a 10 saturated sodium hydrogencarbonate solution. The organic layer was extracted with 10 mL of ethyl acetate, washed with brine, and dried over anhydrous magnesium sulfate. The anhydrous magnesium sulfate was removed by filtration, and the organic layer was concentrated to dryness, and the residue was triturated with a mixed solution consisting of ethyl acetate:n-heptane = 1:1. The obtained 15 solid was filtrated, washed with diethyl ether, and dried under aeration to give 85 mg of a target product. Yield: 87%. 'H-NMR (CD 3 0D) 8 (ppm): 3.01 (3H, s), 3.05 (3H, s), 3.13 (3H, s), 3.83 (3H, s), 3.99 (3H, s), 7.11 (1H, s), 7.27 (lH, s), 7.52 (1H, ddd, J = 1.6, 1.6, 8.0 Hz), 7.57 (2H, d, J = 8.4 Hz), 7.58 (1H, t, J = 8.4 Hz), 7.81 (1H, ddd, J = 1.6, 2.0, 8.0 Hz), 20 8.04 (2H, d, J = 8.4 Hz), 8.19 (1H, t, J = 2.0 Hz). [0049] The following compounds of production examples 4 to 10 were synthesized by methods similar to production example 3, using the compound of production example 2 as a starting substance, and also using the corresponding alcohol or amine. 25 [0050] Production example 4 21 FP08-0370-00 Synthesis of ethyl N-[3-(6,7-dimethoxy-2-methylaminoguinazolin-4 yl)phenyllterephthalamic acid [Formula 11] 0 00 HO CH3
H
3 CO N KI N . CH 3
H
3 CO N N H 5 'H-NMR (DMSO-d) 5 (ppm): 1.33 (314, t, J = 7.2 Hz), 2.84 (3H, d, J = 4.8 Hz), 3.74 (3H, s), 3.91 (3H, s), 4.34 (2H, q, J = 7.2 Hz), 6.99 (114, s), 7.00 (1H, brs), 7.17 (1H, s), 7.47 (1H, d, J = 8.0 Hz), 7.55 (1H, t, J = 8.0 Hz), 7.88 (1H, brd, J = 8.0 Hz), 8.08 (4H, s), 8.20 (1H, brs), 10.61 (1H, s). [0051] Production example 5 10 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoguinazolin-4-yl)phenylI-N' methylterephthalamide [Formula 12] 0
N'CH
3 H H N 0
H
3 CO N
H
3 N NCH3 H 'H-NMR (DMSO-d 6 ) 8 (ppm): 2.81 (3H, d, J = 4.4 Hz), 2.90 (3H, d, J = 5.2 Hz), 15 3.75 (3H, s), 3.93 (3H, s), 6.99 (1H, s), 7.01 (1H, brs), 7.18 (1H, s), 7.46 (1H, d, J = 8.0 Hz), 7.55 (1H, t, J = 8.0 Hz), 7.89 (114, brd, J = 8.0 Hz), 7.96 (2H, d, J = 8.8 Hz), 8.04 (2H, d, J = 8.8 Hz), 8.21 (1H, t, J = 1.6 Hz), 8.59 (11H, br), 10.53 (1H, s). [0052] Production example 6 22 FP08-0370-00 Synthesis of propyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 yl)phenyllterephthalamic acid [Formula 13] 0 0 H N 0
CH
3
H
3 CO N r, ~CH 3
H
3 CO N N H 5 'H-NMR (DMSO-d 6 ) 8 (ppm): 0.99 (3H, t, J 7.6 Hz), 1.76 (2H, m), 2.90 (3H, d, J = 5.2 Hz), 3.76 (3H, s), 3.93 (3H, s), 4.28 (2H, t, J = 6.8 Hz), 7.01 (1H, s), 7.03 (1H, brs), 7.19 (1H, s), 7.49 (1H, d, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.90 (IH, brd, J = 8.0 Hz), 8.11.(4H, s), 8.22 (1H, brs), 10.65 (1H, s). [0053] Production example 7 10 Synthesis of isopropyl N-r3-(6,7-dimethoxy-2-methylaminoquinazolin-4 yl)phenv ljterephthalamic acid [Formula 14] 0 0 HI N
H
3 C
CH
3 OH 0
H
3 CO N
H
3 CO N N'H3 H 'H-NMR (DMSO-d,) 8 (ppm): 1.35 (6H, d, J = 6.4 Hz), 2.90 (3H, d, J = 5.2 Hz), 15 3.76 (3H, s), 3.93 (3H, s), 5.18 (1H, m), 7.01 (IH, s), 7.03 (1H, brs), 7.19 (1H, s), 7.49 (1H, d, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.91 (1H, brd, J = 8.0 Hz), 8.09 (4H, s), 8.22 (1H, brs), 10.65 (1H, s). 23 FP08-0370-00 [0054] Production example 8 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yll-N' ethylterephthalamide [Formula 15] 0 N-CH3 H H N
H
3 CO , H3CO N NCH 3 5 H 'H-NMR (DMSO-d 6 ) 6 (ppm): 1.15 (3H, t, J = 7.2 Hz), 2.91 (3H, d, J = 4.8 Hz), 3.32 (2H, m), 3.76 (3H, s), 3.94 (3H, s), 7.01 (1H, s), 7.03 (1H, brs), 7.19 (1H, s), 7.48 (1H, d, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.91 (1H, brd, J = 8.0 Hz), 7.98 (2H, d, J = 8.4 Hz), 8.06 (2H, d, J= 8.4 Hz), 8.22 (1H, brs), 8.64 (1H, t, J = 5.6 Hz), 10 10.55 (1H, s). [0055] Production example 9 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N' propylterephthalamide [Formula 16] 0 NH H N 0
CH
3
H
3 CO ,
H
3 CO N NCH3 15 H 'H-NMR (DMSO-d 6 ) 8 (ppm): 0.91 (3H, t, J = 7.6 Hz), 1.56 (2H, m), 2.91 (3H, d, J = 4.8 Hz), 3.25 (2H, q, J = 6.0 Hz), 3.76 (3H, s), 3.94 (3H, s), 7.01 (1H, s), 7.02 (1H, brs), 7.19 (IH, s), 7.48 (1H, d, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.91 (1H, brd, J = 8.0 Hz), 7.98 (2H, d, J = 8.4 Hz), 8.06 (2H, d, J = 8.4 Hz), 8.22 (1H, brs), 24 FP08-0370-00 8.62 (1H, t, J = 6.0 Hz), 10.55 (1H, s). [0056] Production example 10 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N' isopropylterephthalamide 5 [Formula 17] 0 NH
H
3 C CH 3 0
H
3 CO N -1: i! 'CH 3
H
3 CO N N H 'H-NMR (DMSO-d,) 8 (ppm): 1.19 (6H, d, J = 6.8 Hz), 2.91 (3H, d, J 4.8 Hz), 3.76 (3H, s), 3.94 (3H, s), 4.12 (1H, m), 7.01 (1H, s), 7.02 (1H, brs), 7.19 (1H, s), 7.48 (1H, d, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.92 (1H, brd, J = 8.0 Hz), 7.98 10 (2H, d, J = 8.4 Hz), 8.05 (2H, d, J = 8.4 Hz), 8.22 (1H, brs), 8.34 (1H, d, J = 7.6 Hz), 10.55 (1H, s). [0057] Production example 11 Synthesis of methyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 yl)phenyllisophthalic acid 15 [Formula 18] H O 0 0
H
3 CO o N :. OH 3
H
3 CO N N' H A mixture of 2.00 g (6.44 mmol) of 3-(2-chloro-6,7-dimethoxyquinazolin 4-yl)phenylamine, 1.75 g (9.71 mmol) of isophthalic acid monomethyl ester, 2.7 mL of triethylamine, 1.00 g of I-hydroxybenzotriazole hydrate, and 2.00 g of WSC 20 hydrochloride, was suspended in 15 mL of dimethylfornamide, followed by 25 FP08-0370-00 stirring at room temperature overnight. The reaction mixture was poured into water, and extracted with ethyl acetate. The organic layer was washed with brine, and dried over magnesium sulfate. After filtration, the residue obtained by solvent distillation under reduced pressure was then subjected to silica gel column 5 chromatography (ethyl acetate-heptane). Thereafter, a solid precipitated with ethyl acetate-hexane was collected by filtration, and dried under aeration to give 2.65 g of the titled compound (yield: 87%). 'H-NMR (DMSO-d 6 ) 8 (ppm): 2.91 (3H, d, J = 4.8 Hz), 3.76 (3H, s), 3.92 (3H, s), 3.93 (3H, s), 7.01 (1H, s), 7.02 (1H, brs), 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 10 7.57 (1H, t, J = 8.0 Hz), 7.72 (1H, t, J = 8.0 Hz), 7.92 (lH, brd, J = 8.0 Hz), 8.17 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J= 1.6 Hz), 8.26 (1H, brd, J= 8.0 Hz), 8.56 (1H, t, J= 1.6 Hz), 10.67 (IH, s). [0058] Production example 12 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 15 yl)phenyllisophthalic acid [Formula 19] H N OH o 0
H
3 CO N
CH
3
H
3 CO N N H To a solution of 2.49 g (5.27 mmol) of the compound of production example 11 as obtained above dissolved in a mixed solvent consisting of 40 mL of 20 tetrahydrofuran and 40 mL of ethanol was added 15 mL of a 1 N sodium hydroxide aqueous solution, followed by stirring at room temperature overnight. The reaction mixture was neutralized with 15 mL of 1 N hydrochloric acid, and 60 mL of water was added thereto. The precipitated solid was collected by filtration, and dried under hot air to give 3.31 g of the titled compound. 25 'H-NMR (DMSO-d 6 ) 8 (ppm): 2.91 (3H, d, J = 4.8 Hz), 3.76 (3H, s), 3.94 (3H, s), 7.01 (1H, s), 7.02 (1H, brs), 7.20 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J 26 FP08-0370-00 = 8.0 Hz), 7.69 (1H, t, J= 8.0 Hz), 7.92 (1H, brd, J = 8.0 Hz), 8.15 (1H, brd, J= 8.0 Hz), 8.22 (1H, brd, J = 8.0 Hz), 8.23 (1H, t, J = 1.6 Hz), 8.56 (1H, t, J = 1.6 Hz), 10.65 (1H, s). [0059] The following compounds of production examples 13 to 19 were 5 synthesized by methods similar to production example 3, using the compound of the aforementioned production example 12 as a starting substance, and also using the corresponding alcohol or amine. [0060] Production example 13 Synthesis of ethyl N-[3-(6.7-dimethoxy-2-methylaminoquinazolin-4 10 yl)phenyllisophthalic acid [Formula 20] H N 01 O .CH 3 O 0
H
3 CO o NN
H
3 CO N N'H3 H 'H-NMR (DMSO-dt 6 ) 8 (ppm): 1.36 (3H, t, J = 7.2 Hz), 2.91 (3H, d, J = 4.8 Hz), 3.76 (3H, s), 3.93 (3H, s), 4.38 (2H, q, J = 7.2 Hz), 7.01 (1H, s), 7.02 (1H, brs), 15 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.71 (1H, t, J = 8.0 Hz), 7.92 (1H, brd, J = 8.0 Hz), 8.17 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J = 1.6 Hz), 8.25 (IH, brd, J = 8.0 Hz), 8.54 (1H, t, J = 1.6 Hz), 10.67 (1H, s). [0061] Production example 14 Synthesis of propyl N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4 20 yl)phenyllisophthalic acid [Formula 21] 27 FP08-0370-00 H N O CH3 0 O
H
3 CO N ~KCH3
H
3 CO N N' H 'H-NMR (DMSO-d 6 ) 8 (ppm): 0.99 (3H, t, J = 7.2 Hlz), 1.76 (2H, qt, J 7.2, 6.8 Hz), 2.91 (3H, d, J = 4.4 Hz), 3.76 (3H, s), 3.93 (3H, s), 4.29 (2H, t, J = 6.8 Hz), 7.01 (1H, s), 7.02 (IH, brs), 7.19 (1H, s), 7.49 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J 5 = 8.0 Hz), 7.72 (1H, t, J = 8.0 Hz), 7.91 (1H, brd, J = 8.0 Hz), 8.18 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J = 1.6 Hz), 8.25 (1H, brd, J = 8.0 Hz), 8.54 (1H, t, J = 1.6 Hz), 10.67 (1H, s). [00621 Production example 15 Synthesis of isopropyl N-[3-(6.7-dimethoxy-2-methylaminoquinazolin-4 10 yl)phenyllisophthalic acid [Formula 22] H N OIr 0 CH 3 0 0 CH 3
H
3 CO '1 N
H
3 CO N N'CH3 H 'H-NMR (DMSO-d,) 8 (ppm): 1.36 (6H, d, J = 6.4 Hz), 2.91 (3H, d, J 4.8 Hz), 3.76 (3H, s), 3.93 (3H, s), 5.19 (1H, septet, J = 6.4 Hz), 7.01 (IH, s), 7.02 (1H, brs), 15 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.71 (1H, t, J = 8.0 Hz), 7.91 (1H, brd, J = 8.0 Hz), 8.15 (1H, brd, J = 8.0 Hz), 8.21 (1H, t, J = 1.6 Hz), 8.24 (1H, brd, J = 8.0 Hz), 8.52 (1H, t, J = 1.6 Hz), 10.67 (1H, s). [0063] Production example 16 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N' 20 methylisophthalamide [Formula 23] 28 FP08-0370-00 H IH N N, NCH3 O O
H
3 CO s'N ~kr
CH
3 H3CO Nl N' H 'H-NMR (DMSO-de) S (ppm): 2.82 (3H, d, J = 4.4 Hz), 2.91 (3H, d, J = 4.8 Hz), 3.76 (3H, s), 3.93 (3H, s), 7.01 (1H, s), 7.02 (1H, brs), 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.64 (1H, t, J = 8.0 Hz), 7.91 (1H, brd, J = 8.0 5 Hz), 8.02 (1H, brd, J = 8.0 Hz), 8.10 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J = 1.6 Hz), 8.42 (1H, t, J = 1.6 Hz), 8.60 (1H, brq, J = 4.8 Hz), 10.58 (1H, s). [0064] Production example 17 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N' ethylisophthalamide 10 [Formula 24] H H N "-( r N CH 3 O 0
H
3 CO sN 1 KCH 3
H
3 CO N N' H 'H-NMR (DMSO-d 6 ) 8 (ppm): 1.15 (3H, t, J = 7.2 Hz), 2.91 (3H, d, J = 4.4 Hz), 3.33 (2H, q, J = 7.2 Hz), 3.76 (3H, s), 3.93 (3H, s), 7.01 (1H, s), 7.02 (1H, brs), 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.63 (1H, t, J = 8.0 15 Hz), 7.92 (1H, brd, J = 8.0 Hz), 8.03 (1H, brd, J = 8.0 Hz), 8.09 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J = 1.6 Hz), 8.42 (1H, t, J = 1.6 Hz), 8.63 (1H, brt, J = 5.4 Hz), 10.58 (1H, s). [0065] Production example 18 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N' 20 propylisophthalamide [Formula 25] 29 FP08-0370-00 N N -"CH3 0 0
H
3 CO ,'N
H
3 CO N .H3 H 'H-NMR (DMSO-d,) 8 (ppm): 0.91 (3H, t, J = 7.2 Hz), 1.56 (2H, qt, J 7.2, 6.4 Hz), 2.91 (3H, d, J = 4.4 Hz), 3.25 (2H, dt, J = 6.4, 5.4 Hz), 3.76 (3H, s), 3.93 (3H, s), 7.01 (1H, s), 7.02 (1H, brs), 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (LH, t, 5 J = 8.0 Hz), 7.63 (1H, t, J = 8.0 Hz), 7.92 (1H, brd, J = 8.0 Hz), 8.04 (11H, brd, J = 8.0 Hz), 8.09 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J = 1.6 Hz), 8.42 (1H, t, J = 1.6 Hz), 8.62 (1H, brt, J = 5.4 Hz), 10.59 (1H, s). [0066] Production example 19 Synthesis of N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyll-N' 10 isopropylisophthalamide [Formula 26] N N cii CH3 0 0 CH 3 H3CO sN
H
3 CO N' N.CH3 H 'H-NMR (DMSO-d 6 ) 8 (ppm): 1.19 (6H, d, J= 6.4 Hz), 2.91 (3H, d, J = 4.4 Hz), 3.76 (3H, s), 3.93 (3H, s), 4.13 (1H, septet, J = 6.4 Hz), 7.01 (1H, s), 7.02 (1H, brs), 15 7.19 (1H, s), 7.48 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.62 (1H, t, J = 8.0 Hz), 7.93 (1H, brd, J = 8.0 Hz), 8.04 (1H, brd, J = 8.0 Hz), 8.08 (1H, brd, J = 8.0 Hz), 8.22 (1H, t, J= 1.6 Hz), 8.40 (1H, brd), 8.41 (1H, t, J = 1.6 Hz), 10.59 (1H, s). [0067] Production example 20 N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyllterephthalamic acid 2 20 methoxyethyl ester [Formula 27] 30 FP08-0370-00 0 OMOCH3 N 0
H
3 CO N NCH3 H A mixture consisting of 55 mg (0.11 mmol) of N-[3-(6,7-dimethoxy-2 methylaminoquinazolin-4-yl)phenyl]terephthalamic acid hydrochloride, 40 p.L (0.51 mmol) of 2-methoxyethanol, 47 pL of triethylamine, 17 mg of 1 5 hydroxybenzotriazole hydrate, and 35 mg of WSC hydrochloride, was suspended in 2 mL of dimethylformamide, followed by stirring at room temperature overnight. The reaction mixture was poured into water, and extracted with ethyl acetate. The organic layer was washed with brine, and was then dried over magnesium sulfate. The residue obtained by filtration and solvent distillation under reduced pressure 10 was subjected to silica gel column chromatography (ethyl acetate-heptane). Thereafter, a solid precipitated with ethyl acetate-hexane was collected by filtration, and was dried under aeration to give 40 mg of the titled compound (yield: 70%). 'H-NMR (DMSO-d 6 ) 8 (ppm): 2.91 (3H, d, J = 4.8 Hz), 3.32 (3H, s), 3.69 (2H, m), 3.76 (3H, s), 3.93 (3H, s), 4.45 (2H, m), 7.01 (1H, s), 7.03 (IH, brs), 7.19 (1H, s), 15 7.49 (1H, brd, J = 7.6 Hz), 7.57 (1H, t, J = 7.6 Hz), 7.90 (lH, brd, J = 7.6 Hz), 8.11 (4H, s), 8.12 (1H, t, J = 1.8 Hz), 10.65 (1H, s). [0068] Production example 21 N-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyllisophthalamic acid 2 methoxyethyl ester 20 [Formula 28] H SN N 0 0
H
3 CO N H3CO N ),N 'CH, H The titled compound was obtained by a method that was equivalent to a method similar to production example 3 using the compound of production 31 FP08-0370-00 example 12 as a starting substance and also using 2-methoxyethanol. 'H-NMR (DMSO-de 6 ) 8 (ppm): 2.91 (3H, d, J = 4.8 Hz), 3.32 (3H, s), 3.69 (2H, m), 3.76 (3H, s), 3.93 (3H, s), 4.46 (2H, m), 7.01 (1H, s), 7.03 (1H, brs), 7.19 (1H, s), 7.49 (1H, brd, J = 8.0 Hz), 7.57 (1H, t, J = 8.0 Hz), 7.73 (1H, t, J = 8.0 Hz), 7.92 5 (1H, brd, J = 8.0 Hz), 8.17 (1H, dt, J = 8.0, 1.6 Hz), 8.22 (1H, t, J = 1.6 Hz), 8.26 (1H, dt, J = 8.0, 1.6 Hz), 8.54 (1H, t, J= 1.6 Hz), 10.68 (1H, s). [0069] Production example 22 Synthesis of t-butyl [3-(2-chloro-6,7-dimethoxyquinazolin-4-yl)phenyllcarbamate [Formula 29]
CH
3 O CH 3
CH
3
H
3 CO N 10
H
3 CO N CI To a mixture of 1.00 g (3.86 mmol) of 2,4-dichloro-6,7 dimethoxyquinazoline, 1.14 g (4.63 mmol) of 3-(N-t-butoxycarbonylamino)phenyl borate, tetrahydrofuran (25 mL), and 2 M sodium carbonate aqueous solution (5 mL) were added palladium acetate (8.84 mg) and 1,1' 15 bis(diphenylphosphino)ferrocene (21.4 mg) in this order, and the mixture was stirred at 60*C for 6.5 hours under a nitrogen atmosphere. The reaction solution was allowed to cool, and ethyl acetate (25 mL) and 5% w/w sodium chloride solution (20 mL) were added to extract the organic layer. The organic layer was washed twice with 5% w/w sodium chloride solution (20 mL) and then 20 concentrated under reduced pressure. To the concentration residue were added ethyl acetate (1 mL) and 2-propanol (4 mL), and the mixture was suspended by stirring at 40*C for 0.5 hours. The suspension was cooled, and the precipitated crystals were collected by filtration and dried to give 1.48 g of a target product (yield: 91.5%, HPLC purity: 99.02%). 25 'H-NMR (CDCl 3 ) S (ppm): 1.52 (9H, s), 3.97 (3H, s), 4.07 (3H, s), 6.62 (1H, br), 7.33 (1H, s), 7.38-7.43 (1H, m), 7.48-7.53 (3H, m), 8.00 (1H, br). ESI MS: mL/z 438 (M+Na)*. [0070] Production example 3 32 FP08-0370-00 Synthesis of t-butyl {3-[6,7-dimethoxy-2-(methylamino)quinazolin-4 yllphenyllcarbamate [Formula 30] H H N 0 CH 3 i'g~ CH 3
CH
3
H
3 CO N ):N% N.llN.CH3
H
3 CO N NH H 5 In a SUS autoclave were placed 420 mg (1.00 mmol) of t-butyl [3-(2 chloro-6,7-dimethoxyquinazolin-4-yl)phenyl]carbamate, tetrahydrofuran (2.5 mL), and 2-propanol (1.25 mL), to this mixture was added a methanol solution (2.5 mL) of 40% methylamine, and the mixture was stirred at 90*C for 8 hours. The reaction mixture was allowed to cool and then poured into a mixed solution of ethyl acetate 10 (40 mL), tetrahydrofuran (40 mL), and 5% w/w sodium chloride solution (50 mL) to extract the organic layer. The organic layer was washed with 5% w/w sodium chloride solution (50 mL) and then concentrated under reduced pressure. To the concentration residue was added t-butyl methyl ether (2.1 mL), and the mixture was crystallized with a spatula and then stirred at room temperature for 3 hours. 15 The precipitated crystals were collected by filtration and dried to give 348 mg of a target product (yield: 83.8%, HPLC purity: 98.70%). 'H-NMR (CDCl 3 ) 8 (ppm): 1.52 (9H, s), 3.12 (3H, d, J=5.2 Hz), 3.85 (3H, s), 4.03 (3H, s), 5.11 (IH, brd, J=5.2 Hz), 6.59 (1H, br), 7.07 (1H, s), 7.19 (1H, s), 7.36 7.48 (3H, m), 7.80 (1H, br). ESI MS: m/z 433 (M+Na)*. 20 [0071] Production example 24 Synthesis of [4-(3-aminophenyl)-6,7-dimethoxyquinazolin-2-yllmethylamine [Formula 31]
NH
2
H
3 CO N H3CO N N.CH3 H Under a nitrogen atmosphere, 100 mg (0.24 mmol) of t-butyl {3-[6,7 33 FP08-0370-00 dimethoxy-2-(methylamino)quinazolin-4-yl]phenyl}carbamate was suspended in dichloromethane (1 mL), to the suspension was dropwise added trifluoroacetic acid (0.2 mL) while cooling to 0*C, the mixture was stirred at the same temperature for 1 hour followed by stirring at room temperature for 6 hours. While cooling with 5 ice water, 0.5 N aqueous sodium hydroxide solution (5.94 mL) was added dropwise, and into the reaction mixture were poured ethyl acetate (10 mL), tetrahydrofuran (10 mL), and 5% w/w sodium chloride solution (20 mL) to extract the organic layer. The organic layer was washed twice with 5% w/w sodium chloride solution (20 mL) and then concentrated under reduced pressure. To the concentration residue 10 was added t-butyl methyl ether (0.6 mL), and the mixture was crystallized with a spatula and stirred at room temperature for 4 hours. The precipitated crystals were collected by filtration and dried to give 66.1 mg of a target product (yield: 87.2%, HPLC purity: 98.27%). 'H-NMR (CDCl 3 ) 8 (ppm): 3.12 (3H, d, J=5.2 Hz), 3.80 (2H, brs), 3.82 (3H, s), 15 4.03 (3H, s), 5.30 (1H, br), 6.83 (1H, dd, J=1.6, 8.0 Hz), 6.99 (1H, t, J=1.6 Hz), 7.04 (1H, brd, J=8.0 Hz), 7.07 (1H, s), 7.15 (1H, s), 7.30 (1H, t, J=8.0 Hz). [0072] Production example 25 A method for producing anhydrous crystals 1 of methyl N-[3-(6,7-dimethoxy-2 methylaminoquinazolin-4-yl)phenyllterephthalamic acid 20 A suspension consisting of 10.00 g (55.51 mmol) of monomethyl terephthalate and 90 mL of 1,2-dimethoxyethane was stirred, while it is cooled in a cold bath at 10*C. To the suspension were added 2.0 mL of N,N dimethylformamide and 6.61 g (52.75 mmol) of thionyl chloride in this order. The suspension was stirred under heating at 60*C to 65*C for 1 hour, and allowed to 25 cool. Thereafter, the suspension was further stirred while it was cooled in an ice bath. Subsequently, 6.83 g (52.82 mmol) of diisopropylethylamine was added dropwise to the mixture. Subsequently, the reaction mixture was stirred at room temperature. Thirty minutes after the internal temperature had reached 20*C, stirring was terminated. The reaction mixture was placed in a 200-mL eggplant 30 flask, followed by measurement to yield 109.49 g of a mixed solution consisting of [monomethyl terephthalate chloride/diisopropylethylamine] (the content of monomethyl terephthalate chloride: 8.89 g) as a slight tannish solution. Subsequently, a suspension consisting of 9.50 g (30.00 mmol) of [4-(3 34 FP08-0370-00 aminophenyl)-6,7-dimethoxyquinazolin-2-yl]methylamine and 380 mL of tetrahydrofuran was stirred, while it was cooled at 0*C. To the suspension was added dropwise over 1 hour, 80.71 g of the above mixed solution consisting of [monomethyl terephthalate chloride/diisopropylethylamine] (the content of 5 monomethyl terephthalate chloride: 6.55 g; 33.00 mmol). The mixture was then stirred at 0*C for 11 hours. Thereafter, 190 mL of ethyl acetate was added to the reaction mixture while cooling at 0*C, and 380 g of a 5% sodium hydrogencarbonate solution was then added dropwise thereto. The reaction mixture was transferred into a separatory funnel, and 190 mL of ethyl acetate was 10 added. After extraction, the organic layer was separated, and washed with 190 g of a 5% sodium chloride solution and 190 mL of water (twice) in this order. The organic layer was concentrated under reduced pressure at 40*C. To the residue was added 143 mL of methanol, and the mixture was stirred while heating to 40 0 C. Thirty-three minutes after initiation of stirring, the temperature of an oil bath was 15 set at 75 0 C. Thereafter, 30 minutes after the internal temperature had exceeded 60*C, the temperature of the oil bath was set at 50 0 C. When the internal temperature was decreased to 55 0 C, 143 mL of 2-propanol was added dropwise thereto. Subsequently, the internal temperature was gradually cooled to 27.3 0 C, and the mixture was then stirred at 20"C for 17 hours. The precipitated crystals 20 were subjected to vacuum filtration, and the resultant was washed with a mixed solution consisting of 14.3 mL of methanol and 14.3 mL of 2-propanol. The resultant was aspirated with a vacuum line for 10 minutes for deliquoring to give 15.72 g of a crude target product (wet body; the content of a product of interest: 13.31 g) as pale yellow crystals (yield: 93.9%). 25 A suspension consisting of 15.48 g of the crude product of interest (wet body) (the content of the product of interest: 13.11 g; 27.00 mmol) and 40 mL of dimethyl sulfoxide was stirred under heating at 60*C, and the crystals were dissolved. The obtained solution was subjected to clarifying filtration, and washed with 10 mL of dimethyl sulfoxide. The filtrate was transferred into a 1,000-mL 30 four-necked glass vessel, which had previously been heated with a 60*C hot water jacket, and the residue was washed with 10 mL of dimethyl sulfoxide. The mixture was then stirred under heating at 60*C. Thereafter, 119 mL of 2-propanol was added dropwise to this solution, and 49.3 mg of seed crystals of the product of 35 FP08-0370-00 interest was placed in the mixture. Thereafter, 60 mL of 2-propanol was further added dropwise to the mixture. This suspension was stirred at 60"C for 2 hours, the temperature of the jacket was set at 80*C, and the suspension was continuously stirred under heating for 16.5 hours. Subsequently, 120 mL of 2-propanol was 5 added dropwise to the suspension, and 3 hours later, 362 mL of 2-propanol was further added dropwise thereto. Thereafter, the mixture was gradually cooled to 20*C (10*C/h), and it was then stirred at the same temperature. Fifty nine point five hours later, the precipitated crystals were collected by filtration, and the crystals were washed with a mixed solution consisting of 2.6 mL of dimethyl 10 sulfoxide and 24 mL of 2-propanol. The crystals were further washed with 40 mL of 2-propanol, and were then aspirated with a vacuum line for deliquoring. The obtained crystals were dried under reduced pressure to give 9.84 g of a target product as yellow crystals (yield: 73.7%). The measurement of powder X-ray diffraction pattern of the obtained 15 crystals was carried out according to the powder X-ray diffraction measurement method described in General Tests in the Japanese Pharmacopoeia, under the following conditions. (Apparatus) Rigaku X-ray DTA System: RINT-2000 (manufactured by Rigaku Corporation) 20 (Operation method) A sample was ground in an agate mortar, and then sampled on a copper board. Thereafter, measurement was carried out under the following conditions. X-ray used: CuKa ray Tube voltage: 40 kV 25 Tube current: 200 mA Divergent slit: 1/2 deg Receiving slit: 0.3 mm Scattering slit: 1/2 deg Scanning rate: 2*/min 30 Scanning step: 0.020 Scanning range (20): 5" to 400 The powder X-ray diffraction patterns of the obtained crystals is shown in Figure 3. The major peaks of diffraction angles (20) are 8.2*, 16.5* and 24.50. 36 FP08-0370-00 dropwise 16.70 kg of "monomethyl terephthalate chloride/N,N diisopropylethylamine" solution (monomethyl terephthalate chloride content: 1.40 kg, 7.03 mol) over 1 hour and 26 minutes, and the container was washed with 1.40 L of 1,2-dimethoxyethane. The mixture was stirred at 0*C for 13 hours and 4 5 minutes. Under cooling at 0*C, 36.5 kg of ethyl acetate was added to the reaction mixture and then 80.1 kg of a 5% aqueous solution of sodium hydrogencarbonate was added dropwise, and the mixture was stirred at 20*C for 1 hour and 10 minutes. Then, 37.3 kg of ethyl acetate was added into the mixture, the mixture was stirred, and the water layer was separated. The organic layer was washed with 40.0 kg of a 10 5% aqueous solution of sodium chloride, 40.2 kg of water, and 40.1 kg of water in this order. The organic layer was concentrated under reduced pressure at a jacket temperature of 40*C, 23.70 kg of methanol was added to the residue, and stirred for 1 hour and 1 minute while being heated to 60 to 66*C. 23.60 kg of 2-propanol was added dropwise to the suspension over 1 hour while stirring the suspension at a 15 jacket temperature of 50*C. Then, the suspension was cooled at a cooling rate of 1 0 C/hour and stirred at 20*C for 12 hours and 23 minutes. The precipitated crystals were filtered, rinsed with a mixed solution of 3.00 L of methanol and 3.00 L of 2-propanol and 6.00 L of 2-propanol in this order to give 5.52 kg of a crude product (content of the target compound: 2.57 kg, 5.44 mol) as pale yellow crystals 20 (yield: 85.3%). In a nitrogen atmosphere, a suspension of 5.398 kg of the crude product (content of the target compound: 2.518 kg, 5.33 mol) in 8.01 L of dimethyl sulfoxide was stirred under heating at 60 to 70*C, and the crystals were dissolved. The solution was filtered, and rinsed with 2.00 L of dimethyl sulfoxide. The 25 filtrate was transferred into a 210 L reaction vessel having been heated at 60*C and the container was washed with 2.01 L of dimethyl sulfoxide. To the solution, 18.9 kg of 2-propanol was added dropwise over 40 minutes, 15.02 g of crystals of the target compound was seeded, and 9.44 kg of 2-propanol was added dropwise over 57 minutes. After stirring the suspension at 60*C for 1 hour and 30 minutes, the 30 jacket temperature was set at 80*C and the stirring was continued for 37 hours and 24 minutes. Then, 56.6 kg of 2-propanol was added dropwise to the suspension over 2 hours and 8 minutes, the mixture was cooled to 20*C at a cooling rate of 10*C/hour and stirred at the same temperature for 65 hours and 50 minutes. The 38 FP08-0370-00 precipitated crystals were filtered, rinsed with a mixed solution of 534 mL of dimethyl sulfoxide and 4.81 L of 2-propanol and 8.01 L of 2-propanol in this order. The crystals were dried under reduced pressure at 50*C to give 2.30 kg of the target product as yellow crystals (yield 90.8%). 5 [0074] Example 1 An ointment containing methyl N-[3-(6,7-dimethoxy-2 methylaminoquinazolin-4-yl)phenyl]terephthalamic acid anhydride (hereinafter referred to as Compound A) of Production example 25 was manufactured according to the prescription shown in Table 1. First, Compound A was dissolved 10 by heating at 80*C together with polyethylene glycol 400 and dl-a-tocopherol (Solution D. Meanwhile, white petrolatum, polyethylene glycol 4000, isopropyl myristate, glyceryl monostearate, stearic acid, and polyoxyethylene hydrogenated castor oil 40 were dissolved at 80*C and mixed by stirring. Solution I was added to the ointment base, and mixed by stirring at 80*C for 3 minutes. Then the mixture 15 was cooled to 32*C with stirring in order to produce an ointment containing 0.01% by weight of Compound A. [0075] [Table 1] Ingredient Amount (% by weight) Compound A 0. 01 White petrolatum 48. 9 Polyethylene glycol 400 20 Isopropyl myristate 10 Polyethylene glycol 4000 10 Glyceryl monostearate 5 Stearic acid 5 Polyoxyethylene hydrogenated castor oil 40 1 dl-a-Tocopherol 0. 1 [0076] Example 2 20 An ointment containing 0.01% by weight of Compound A was manufactured according to the prescription shown in Table 2 in the same manner as in Example 1. [0077] [Table 2] 39 FP08-0370-00 Ingredient Amount (% by weight) Compound A 0. 01 White petrolatum 50. 9 Polyethylene glycol 400 20 Isopropyl myristate 10 Polyethylene glycol 4000 10 Glyceryl monostearate 8 Polyoxyethylene hydrogenated castor oil 60 1 dl-a-Tocopherol 0. 1 [0078] Comparative examples 1 to 5 An ointment containing 0.01% by weight of Compound A was manufactured according to the prescription shown in Table 3 in the same manner as 5 in Example 1. [0079] [Table 3] Amount (% by weight) Comp. Ex.1 Comp. Ex.2 Comp. Ex.3 Comp. Ex.4 Comp. Ex.5 Compound A 0.01 0.01 0.01 0.01 0.01 White petrolatum 59. 9 59. 9 59. 9 59. 9 59. 9 Polyethylene glycol 400 20 20 20 10 10 Isopropyl myristate - 5 10 10 10 Paraffin 5 5 5 5 5 White beeswax 5 5 5 10 5 Squalane 10 5 - - Cetanol - - - 5 Polyethylene glycol 4000 - - - - 10 dl-a-Tocopherol 0. 1 0. 1 0. 1 0. 1 0. 1 [0080] Test example I The ointments of Examples 1 and 2 and Comparative examples 1 to 5 were 10 filled in clear glass bottles and stored at room temperature for 1 week, and then the ointments were visually inspected for the presence or absence of bleeding of the liquid ingredients from the formulation (A, not bled; B slightly bled; C, bled). Furthermore, an appropriate amount of each ointment was applied to the back of the hand immediately after production, and usability was evaluated (A, 15 properly hard, B, slightly hard and difficult to apply; C, too hard for practical use). The drug efficacy of each ointment was evaluated in the same manner as in Test examples 2 and 3 (A, markedly effective; B, effective; C, no effect). The results are shown in Tables 4 and 5. [0081] [Table 4] 40 FP08-0370-00 Ex.1I Ex.E 2 Bleeding A A Usability A A Drug efficacy A A [0082] [Table 5] Comp.Ex.1 Comp.Ex.2 Comp.Ex.3 Comp.Ex.4 Comp.Ex.5 Bleeding B C C C C Usability A - Drug efficacy C B A - - Not tested [0083] As a result of each of the above tests, the ointments of Examples 1 and 2 5 were superior in any evaluation item. Although Comparative example 1, which did not contain an absorption enhancer, was excellent in prevention of bleeding and usability, no drug efficacy was observed. Furthermore, Comparative examples 2 and 3, which contained an absorption enhancer instead of part or all of squalane in Comparative example 1, showed drug efficacy depending on the amount of the 10 absorption enhancer mixed, but bleeding of liquid ingredients was observed. Furthermore, Comparative examples 4 and 5, which contained cetanol or polyethylene glycol 4000, also showed the bleeding. [0084] Test example 2 Effect in oxazolone-induced dermatitis model An ointment containing 0.003, 0.01, or 0.03% of Compound A was 15 produced in the same manner as in Example 1. An effect of suppressing dermatitis of this ointment was examined using the mouse model having clinical symptoms of dermatitis. 1) Breeding As test animals, 5-week-old NC/Nga female mice (Japan Charles River 20 Laboratories Japan, Inc.) were used. After 1 week or longer of acclimatization/preliminary breeding, only animals, wherein no abnormal changes were found in a general state, were used for the test. 2) Sensitization and induction of dermatitis Sensitization was carried out by applying 10 paL of an acetone solution 25 (Wako Pure Chemical Industries, Ltd.) that contained 0.3% 4-ethoxymethylene-2 phenyl-2-oxazolin-5-one (hereinafter abbreviated as "oxazolone": Sigma) to each of the left and right pinnas of mice. Induction was carried out by applying 10 paL of 0.3% oxazolone to the left 41 FP08-0370-00 pinna of each mouse, twice in total, on the 5th day after sensitization and 3 days later. Dermatitis was induced, and the mice were divided into groups, such that symptoms in each group became uniform. 3) Application of ointment 5 The aforementioned ointment (approximately 10 mg) was applied to the left pinna of each mouse once daily (2 hours or longer before stimulation with oxazolone on the stimulation days) for 7 consecutive days from the 1st day, when the ointment application was started. An ointment not containing Compound A was applied as a control group. 10 4) Evaluation of ointment Each animal was observed for the condition of dermatitis once daily before application of the drug from the 1st day to the 8th day (no application on the 8th day). Symptoms were evaluated using scores according to the following criteria, the sum of scores of each item was obtained as the score of each individual mouse, 15 and the mean score was calculated for each treatment group. Criteria for each of erythema, abrasion and crust/erosion: 0, no symptom; 1, mild; 2, moderate; 3, severe. 5) Test results The results of dermatitis rated using scores are shown in Figure 1. As a 20 result of the above test, a dose-dependent dermatitis improving effect was observed in the groups given the ointment containing Compound A. [0085] Test example 3 Effect in oxazolone-induced scratching behavior model 1) Breeding As test animals, 5-week-old NC/Nga female mice (Japan SLC, Inc.) were 25 used. For acclimation, the mice passed a preliminary breeding period of 7 days. After 1 week or longer of acclimatization/preliminary breeding, only animals, wherein no changes were found in a general state, were used for the test. 2) Sensitization and induction of dermatitis Sensitization was carried out by applying 20 iL of an acetone solution that 30 contained 0.5% oxazolone to each of the left and right pinnas of mice, which had passed an acclimation/preliminary breeding period. Induction was carried out by applying 10 pL of 0.3% oxazolone to the left pinna of each mouse, 3 times in total, at the 4th day after sensitization, at 2 or 3 42 FP08-0370-00 days after the 4th day after sensitization and at 2 or 3 days after said date. 3) Measurement of scratching behavior Scratching behavior of mice was automatically measured using a pruritus measuring device (Micro Act Device: NeuroScience, Inc.). Mice were anesthetized 5 with diethyl ether (Wako Pure Chemical Industries, Ltd.), and a magnet piece (diameter, 1 mm; length, 3 mm: NeuroScience, Inc.) was subcutaneously inserted into the left hind leg of each mouse by the day before measurement at latest. Oxazolone was applied to induce scratching behavior, then the mice were transferred into a chamber (diameter, 11 cm; height, 18cm) around which a coil is 10 wound, and electric current induced by the movement of the magnet inserted into the leg of the mouse was measured for a certain period of time. A characteristic wave form that reflects such scratching behavior was detected by the pruritus measuring device, and the appearance frequency of the detected wave form was counted as a number of scratching behaviors. 15 4) Administration of ointment The aforementioned ointment (approximately 10 mg) was applied to the left pinna at 4 hours before oxazolone stimulation. The following five test groups were determined: (1) normal group - an ointment not containing Compound A application group in which dermatitis was not induced; (2) control group - an 20 ointment not containing Compound A application group; (3) an ointment containing 0.003% Compound A application group; (4) an ointment containing 0.01% Compound A application group; and (5) an ointment containing 0.03% Compound A application group. The mice were divided into groups, such that the number of scratching behaviors in each group became uniform based on the 25 number of scratching behaviors obtained during the 2nd induction scratching. 5) Evaluation of ointments Evaluation was carried out using the number of scratching behaviors induced by the 3rd application of oxazolone (acetone was applied to the normal group) as an indicator. The number of scratching behaviors was measured at 2 30 hours after application of oxazolone. 6) Test results The results of the number of scratching behaviors are shown in Figure 2. From these results, it was found that the ointments of the present invention 43 C.\Rorb\DC\GD B\2 9923 3_ .DOC-6/10(2010 suppress scratching behavior and also suppress deterioration in cutaneous symptoms caused by such scratching behavior, thereby having an excellent anti- pruritic effect. [0086] Examples 3 to 5 An ointment containing 0.01% by weight of Compound A was manufactured 5 according to the prescription shown in Table 6 in the same manner as in Example 1. Then, the ointments of Examples 3 to 5 were evaluated in the same manner as in Test example 1. The results are shown in Table 6. The ointments of Examples 3 to 5 were superior in the evaluation items of bleeding and usability. [0087] [Table 6] 10 Ingredient Amount (% by weight) Example 3 Example 4 Example 5 Compound A 0.01 0.01 0.01 White petrolatum 55 60 5 Polyethylene glycol 400 20 10 2 Isopropyl myristate 5 10 1 Polyethylene glycol 4000 10 10 1 Glyceryl monostearate 10 5 1 Stearic acid - 5 dl-a-Tocopherol 0. 1 0.1 0. 1 Evaluation item Bleeding A A A Usability A A A [0088] Examples 6 to 9 An ointment containing 0.01% by weight of Compound A was manufactured according to the prescription shown in Table 7 in the same manner as in Example 1. 15 Water was added together with Solution I. Then, the ointments of Examples 6 to 9 were evaluated in the same manner as in Test example 1. The results are shown in Table 7. The ointments of Examples 6 to 9 were superior in the evaluation items of bleeding and usability. [0089] [Table 7] 44 FP08-0370-00 Ingredient Amount (% by weight) Ex.6 Ex.7 Ex.8 Ex.9 Compound A 0.01 0.01 0.01 0.01 White petrolatum 50. 9 50. 4 49. 9 48. 9 Water 0 0. 5 1 2 Polyethylene glycol 400 20 20 20 20 Isopropyl myristate 10 10 10 10 Polyethylene glycol 4000 10 10 10 10 Glyceryl monostearate 8 8 8 8 Polyoxyethylene hydrogenated castor oil 60 1 1 1 1 dl-a-Tocopherol 0. 1 0. 1 0. 1 0. 1 Evaluation item Bleeding A A A A Usability A A A A [0090] Test Example 4 Evaluation of stability of ointments The ointments of Examples 6 to 9 were filled in a 20-mL light-resistant glass bottle and stored at 5*C or 40*C for a predetermined period, and the amount 5 of impurities was measured by HPLC to evaluate the stability of ointments. The results are shown in Table 8 (numerical values are the values (%) obtained by dividing the main impurities peak area by the active ingredient peak area, and ND indicates that impurities could not be detected). Impurities were detected in the ointment of Example 6, to which water was not added and which was stored at 5"C 10 for 2 weeks, but no impurities were detected in the ointment of Example 9, to which 2% water was added and which was stored at 40*C for 1 month. [0091] [Table 8] Example Amount of water Initial 5At 2 wee At mon0C 6 0% ND 0.58 1.49 1. 33 2.01 7 0.5% ND ND 0.62 0.46 1.05 8 1% ND ND ND ND 0.76 9 2% ND ND ND ND ND [0092] The conditions of HPLC were as follows. 15 Detector: Ultraviolet absorption meter (measurement wavelength 249 nm) Column: Stainless tube (inner diameter, 4.6 mm; length, 25 cm) filled with 5 pm octadecylsilylated silica gel for liquid chromatography Column temperature: Constant temperature around 45*C Mobile phase: Mixed solution of water/acetonitrile/70% perchloric acid 20 (120:80:1) 45 C \NRPonb\DCC RBR\299238 I DOC-6/9/21 I0 Flow rate: Approximately 1.5 mL/min Industrial Applicability [0093)] The topical formulation of the present invention can be used as a therapeutic agent for allergic dermatitis such as atopic dermatitis, contact dermatitis and the like. 5 Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. 10 The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates. 46

Claims (15)

1. A topical formulation, comprising a compound represented by the formula (I), salt thereof, or hydrate thereof, a solvent, a base and water: 5 H N R s 0o H 3 CO. ~'N CH, H 3 CO- N N' H wherein R represents hydroxyl, C 1 . 6 alkoxy optionally substituted with C 1 . 6 alkoxy, or amino optionally substituted with C 1 . 6 alkyl. 10
2. The topical formulation according to claim 1, wherein the compound represented by the formula (I) is methyl N-[3-(6,7-dimethoxy-2 methylaminoquinazolin-4-yl)phenyl]terephthalamic acid.
3. The topical formulation according to claim 1 or 2, comprising an absorption enhancer. 15
4. The topical formulation according to any one of claims 1 to 3, comprising a bleeding preventing agent.
5. The topical formulation according to claim 4, comprising two or more types of bleeding preventing agents.
6. The topical formulation according to any one of claims I to 5, wherein the 20 base is one or more types selected from the group consisting of petrolatum, paraffin, liquid paraffin, microcrystalline wax, carunauba wax, and white beeswax.
7. The topical formulation according to any one of claims 1 to 6, wherein the solvent is one or more types selected from the group consisting of polyethylene glycol having a molecular weight of 200 to 600, dipropylene glycol, benzyl alcohol, 25 polyoxyethylene sorbitan fatty acid ester, diethylene glycol monoethyl ether, propylene glycol, polyoxyethylene oleyl ether, polyoxyethylene octyl phenyl ether, polyoxyethylene lauryl ether, polyoxyethylene castor oil, and oleic acid.
8. The topical formulation according to any one of claims 3 to 7, wherein the 47 C.\NRPonbr\DCCRBR\44(92_ I DOC-111121112 absorption enhancer is one or more types selected from the group consisting of isopropyl myristate, ethyl myristate, octyldodecyl myristate, isopropyl palmitate, isostearyl palmitate, isopropyl isostearate, butyl stearate, ethyl oleate, decyl oleate, diisopropyl sebacate, diethyl sebacate, diisopropyl adipate, diethyl adipate, and diethyl 5 phthalate.
9. The topical formulation according to any one of claims 4 to 8, wherein the bleeding preventing agent is one or more types selected from the group consisting of polyethylene glycol having a molecular weight of 1000 to 50000, polyoxyethylene hydrogenated castor oil, stearic acid, oleic acid, sorbitan monostearate, sorbitan 10 monooleate, sorbitan sesquioleate, sorbitan trioleate, and glycerol esters of fatty acids.
10. The topical formulation according to claim 9, wherein the glycerol esters of fatty acids is one or more types selected from the group consisting of glyceryl monostearate, diglyceryl isostearate, and hexaglyceryl polyricinoleate.
11. The topical formulation according to claim 9, wherein the bleeding 15 preventing agent is polyethylene glycol having a molecular weight of 1000 to 50000 and glycerol esters of fatty acids.
12. The topical formulation according to claim 11, wherein the glycerol esters of fatty acids is glyceryl monostearate.
13. The topical formulation according to any one of claims 3 to 12, wherein the 20 topical formulation comprises 10 to 30% by weight of a solvent and 5 to 20% by weight of an absorption enhancer, and the sum of the solvent and absorption enhancer is 20 to 40% by weight based on the total preparation amount, respectively.
14. A method for preventing bleeding of liquid ingredients, comprising mixing polyethylene glycol having a molecular weight of 1000 to 50000 and glycerol esters of 25 fatty acids in a topical formulation according to any one of claims I to 13.
15. A topical formulation according to claim 1 substantially as hereinbefore described with reference to any one of the Examples. 48
AU2008290001A 2007-08-17 2008-08-15 Novel preparation for external use Ceased AU2008290001B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2007212926 2007-08-17
JP2007-212926 2007-08-17
PCT/JP2008/064621 WO2009025239A1 (en) 2007-08-17 2008-08-15 Novel preparation for external use

Publications (2)

Publication Number Publication Date
AU2008290001A1 AU2008290001A1 (en) 2009-02-26
AU2008290001B2 true AU2008290001B2 (en) 2012-08-09

Family

ID=40378144

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2008290001A Ceased AU2008290001B2 (en) 2007-08-17 2008-08-15 Novel preparation for external use

Country Status (10)

Country Link
US (1) US8513269B2 (en)
EP (1) EP2202229B1 (en)
JP (1) JP5060561B2 (en)
KR (1) KR20100042246A (en)
CN (1) CN101687819B (en)
AT (1) ATE549320T1 (en)
AU (1) AU2008290001B2 (en)
CA (1) CA2696793A1 (en)
TW (1) TWI414296B (en)
WO (1) WO2009025239A1 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2678477A1 (en) 2007-02-16 2008-08-21 Eisai R&D Management Co., Ltd. Crystal, amorphous form and salt of methyl n-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid
CN102336717B (en) 2007-08-17 2013-09-18 卫材R&D管理有限公司 Quinazoline derivative
CA2846752A1 (en) * 2011-08-31 2013-03-07 Amakem Nv Biphenylcarboxamides as rock kinase inhibitors
JP6004364B2 (en) * 2011-10-14 2016-10-05 大正製薬株式会社 Topical skin preparation
JP2013147442A (en) * 2012-01-18 2013-08-01 Kose Corp Skin permeation promoter for stearyl glycyrrhetinate
WO2014133020A1 (en) * 2013-02-28 2014-09-04 エーザイ・アール・アンド・ディー・マネジメント株式会社 Agent for preventing and treating respiratory disease
KR20190021309A (en) 2016-06-07 2019-03-05 더마반트 사이언시즈 게엠베하 Topical formulation of PDE-4 inhibitors and methods of use thereof
TWI717658B (en) * 2017-11-20 2021-02-01 大陸商江蘇恒瑞醫藥股份有限公司 Pharmaceutical composition for topical administration and preparation method thereof
CN111683641A (en) * 2017-12-07 2020-09-18 德玛万科学有限公司 Topical ointment formulations of PDE-4 inhibitors and their use in treating skin conditions
WO2019113475A1 (en) * 2017-12-07 2019-06-13 Dermavant Sciences GmbH Topical ointment formulations and their use in treating skin conditions
CA3107702A1 (en) * 2018-08-16 2020-02-20 Dr. Reddy's Laboratories Ltd. Topical oleaginous compositions
WO2021262956A1 (en) * 2020-06-24 2021-12-30 Roivant Sciences Gmbh Topical gel formulations and their use in treating skin conditions

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6352989B1 (en) * 1998-01-26 2002-03-05 Eisai Co., Ltd. Nitrogenous heterocyclic derivatives and medicine thereof

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE150304T1 (en) 1990-09-04 1997-04-15 Fujisawa Pharmaceutical Co OINTMENTS CONTAINING TRICYCLIC COMPOUNDS
JP3466305B2 (en) 1994-12-12 2003-11-10 久光製薬株式会社 Dissolving agent and external preparation containing the dissolving agent
EP0954315A2 (en) 1996-09-13 1999-11-10 Sugen, Inc. Use of quinazoline derivatives for the manufacture of a medicament in the treatment of hyperproliferative skin disorders
AR015966A1 (en) 1997-10-17 2001-05-30 Smithkline Beecham Corp USE OF A PDE4 INHIBITOR COMPOUND FOR THE PREPARATION OF A USEFUL MEDICINAL PRODUCT FOR PRURITE TREATMENT
JP3373838B2 (en) 1999-10-25 2003-02-04 山之内製薬株式会社 Naphthyridine derivative
AR029185A1 (en) 1999-10-25 2003-06-18 Yamanouchi Pharma Co Ltd NAFTIRIDINE DERIVATIVE
AU2003232828B2 (en) 2002-05-28 2010-06-24 Takeda Gmbh Topically applicable pharmaceutical preparation
US20040038958A1 (en) 2002-07-11 2004-02-26 Chris Rundfeldt Topical treatment of skin diseases
JP4387717B2 (en) 2003-07-11 2009-12-24 株式会社資生堂 Water-in-oil type skin preparation for external use
JP2005047909A (en) 2003-07-17 2005-02-24 Santen Pharmaceut Co Ltd Remedy for pruritus containing piperidine derivative as active ingredient
WO2005007161A1 (en) 2003-07-17 2005-01-27 Santen Pharmaceutical Co., Ltd. Remedy for pruritus comprising piperidine derivative as the active ingredient
WO2005082865A1 (en) 2004-02-27 2005-09-09 Astellas Pharma Inc. Fused bicyclic pyrimidine derivative
JP2008137892A (en) 2005-03-04 2008-06-19 Eisai Co Ltd Antipruritic agent
CN101389612B (en) 2006-02-21 2011-09-21 卫材R&D管理有限公司 4-(3-benzoylaminophenyl)-6,7-dimethoxy-2- methylaminoquinazoline derivative
TWI404709B (en) 2006-02-21 2013-08-11 Eisai R&D Man Co Ltd 4-(3-benzamidophenyl) -6,7-dimethoxy-2-methylamine quinazoline derivatives
CA2678477A1 (en) 2007-02-16 2008-08-21 Eisai R&D Management Co., Ltd. Crystal, amorphous form and salt of methyl n-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6352989B1 (en) * 1998-01-26 2002-03-05 Eisai Co., Ltd. Nitrogenous heterocyclic derivatives and medicine thereof

Also Published As

Publication number Publication date
US20110021545A1 (en) 2011-01-27
JP5060561B2 (en) 2012-10-31
TW200922589A (en) 2009-06-01
ATE549320T1 (en) 2012-03-15
TWI414296B (en) 2013-11-11
EP2202229A1 (en) 2010-06-30
CN101687819B (en) 2013-03-20
CN101687819A (en) 2010-03-31
US8513269B2 (en) 2013-08-20
CA2696793A1 (en) 2009-02-26
AU2008290001A1 (en) 2009-02-26
JPWO2009025239A1 (en) 2010-11-25
EP2202229B1 (en) 2012-03-14
KR20100042246A (en) 2010-04-23
WO2009025239A1 (en) 2009-02-26
EP2202229A4 (en) 2010-09-08

Similar Documents

Publication Publication Date Title
AU2008290001B2 (en) Novel preparation for external use
US10357495B2 (en) Topical formulations of PDE-4 inhibitors and their methods of use
EP1992622B1 (en) 4-(3-benzoylaminophenyl)-6,7-dimethoxy-2- methylaminoquinazoline derivative
CA3127686A1 (en) Topical phosphoinositide 3-kinase inhibitors
EP3750883B1 (en) Aminoalkyl compound
ES2368089T3 (en) DERIVATIVES OF 4- (3-BENZOILAMINOFENIL) -6,7-DIMETOXI-2-METHYLAMINOQUINAZOLINE.
KR101878621B1 (en) Prodrug

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired